Cancer Res Treat.  2023 Jul;55(3):814-831. 10.4143/crt.2022.1315.

Genomic Characteristics and the Potential Clinical Implications in Oligometastatic Non–Small Cell Lung Cancer

Affiliations
  • 1Department of Cancer Center, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
  • 2Geneplus-Beijing, Beijing, China
  • 3Chongqing Clinical Research Center for Geriatrics and Gerontology, Chongqing, China

Abstract

Purpose
Oligometastatic non–small cell lung cancer (NSCLC) patients have been increasingly regarded as a distinct group that could benefit from local treatment to achieve a better clinical outcome. However, current definitions of oligometastasis are solely numerical, which are imprecise because of ignoring the biological heterogeneity caused by genomic characteristics. Our study aimed to profile the molecular alterations of oligometastatic NSCLC and elucidate its potential difference from polymetastasis.
Materials and Methods
We performed next-generation sequencing to analyze tumors and paired peripheral blood from 77 oligometastatic and 21 polymetastatic NSCLC patients to reveal their genomic characteristics and assess the genetic heterogeneity.
Results
We found ERBB2, ALK, MLL4, PIK3CB, and TOP2A were mutated at a significantly lower frequency in oligometastasis compared with polymetastasis. EGFR and KEAP1 alterations were mutually exclusive in oligometastatic group. More importantly, oligometastasis has a unique significant enrichment of apoptosis signaling pathway. In contrast to polymetastasis, a highly enriched COSMIC signature 4 and a special mutational process, COSMIC signature 14, were observed in the oligometastatic cohort. According to OncoKB database, 74.03% of oligometastatic NSCLC patients harbored at least one actionable alteration. The median tumor mutation burden of oligometastasis was 5.00 mutations/Mb, which was significantly associated with smoking, DNA damage repair genes, TP53 mutation, SMARCA4 mutation, LRP1B mutation, ABL1 mutation.
Conclusion
Our results shall help redefine oligometastasis beyond simple lesion enumeration that will ultimately improve the selection of patients with real oligometastatic state and optimize personalized cancer therapy for oligometastatic NSCLC.

Keyword

Non-small cell lung cancer; Oligometastatic; Genomic profiling; High-throughput nucleotide sequencing; Tumor mutational burden

Figure

  • Fig. 1 Alteration landscape of 77 oligometastatic (A) and 21 polymetastatic (B) non–small cell lung cancer (NSCLC) patients. The heat map shows top 20 genes across all samples, with genes ranked by mutation frequency. Top bar summarizes the total number of mutations in each patient (columns). Side bar (rows) summarizes the percentage of tumors with mutation in each gene and mutation composition for each gene in the entire cohort. Bottom heat map, group, smoking, sex, and age. Different colors denote different types of mutations and different clinical features. Single nucleotide variations (C), Ti/Tv ratios in oligometastasis (D) and polymetastasis (E), respectively. Differentially mutated genes between oligometastatic and polymetastatic NSCLC (F). The mutual exclusivity and co-occurrence analysis in each group (G, H).

  • Fig. 2 Top 15 enriched pathways by Kyoto Encyclopedia of Genes and Genomes (KEGG) (A) and functional terms by GO enrichment (B) of somatic mutations in oligometastatic and polymetastatic non–small cell lung cancer (NSCLC) patients. Count: the number of mutations enriched in this signaling pathway or functional term. GO, gene ontology; HIF-1, hypoxia inducible factor 1; KEGG, Kyoto Encyclopedia of Genes and Genomes; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of rapamycin; NSCLC, non–small cell lung cancer; PI3K, phosphoinositide 3-kinase; VEGF, vascular endothelial growth factor.

  • Fig. 3 Mutational signatures in oligometastatic and polymetastatic non–small cell lung cancer (NSCLC) patients. (A) Ninety-six subtype classification of the mutational signature of oligometastatic and polymetastatic NSCLC. (B) Histogram of mutation signatures for oligometastatic and polymetastatic NSCLC patients. RSS, residual sum of squares.

  • Fig. 4 Somatic alterations identified by the 1021-panel that are clinically actionable in oligometastatic group. (A) Samples were classified according to their highest level of actionable alterations. (B) Distribution of alteration types. (C) Distribution of levels of actionable alterations. ALK, anaplastic lymphoma kinase; BRAF, B-Raf proto-oncogene, serine/threonine kinase; CDKN2A, cyclin dependent kinase inhibitor 2A; EGFR, epidermal growth factor receptor; ERBB2, Erb-B2 receptor tyrosine kinase 2; PTEN, phosphatase and tensin homolog.

  • Fig. 5 (A) Distribution of tumor mutation burden (TMB) in 42 oligometastatic and nine polymetastatic non–small cell lung cancer patients. Non-parametric test of TMB according to metastatic state (B), smoking status (C), DNA damage repair (DDR) genes (D), TP53 genotype (E), SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily A, member 4 (SMARCA4) genotype (F), LDL receptor related protein 1B (LRP1B) genotype (G), and ABL1 genotype (H).


Reference

References

1. Roach MC, Robinson CG, DeWees TA, Ganachaud J, Przybysz D, Drzymala R, et al. Stereotactic body radiation therapy for central early-stage NSCLC: results of a prospective phase I/II Trial. J Thorac Oncol. 2018; 13:1727–32.
Article
2. Postmus PE, Kerr KM, Oudkerk M, Senan S, Waller DA, Vansteenkiste J, et al. Early and locally advanced non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017; 28:iv1–21.
Article
3. Rusthoven KE, Hammerman SF, Kavanagh BD, Birtwhistle MJ, Stares M, Camidge DR. Is there a role for consolidative stereotactic body radiation therapy following first-line systemic therapy for metastatic lung cancer? A patterns-of-failure analysis. Acta Oncol. 2009; 48:578–83.
4. Hellman S, Weichselbaum RR. Oligometastases. J Clin Oncol. 1995; 13:8–10.
Article
5. Chang JY, Verma V. Optimize local therapy for oligometastatic and oligoprogressive non-small cell lung cancer to enhance survival. J Natl Compr Canc Netw. 2022; 20:531–9.
Article
6. Iyengar P, Wardak Z, Gerber DE, Tumati V, Ahn C, Hughes RS, et al. Consolidative radiotherapy for limited metastatic non-small-cell lung cancer: a phase 2 randomized clinical trial. JAMA Oncol. 2018; 4:e173501.
7. Gomez DR, Tang C, Zhang J, Blumenschein GR Jr, Hernandez M, Lee JJ, et al. Local Consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: long-term results of a multi-institutional, phase II, randomized study. J Clin Oncol. 2019; 37:1558–65.
Article
8. Bauml JM, Mick R, Ciunci C, Aggarwal C, Davis C, Evans T, et al. Pembrolizumab after completion of locally ablative therapy for oligometastatic non-small cell lung cancer: a phase 2 trial. JAMA Oncol. 2019; 5:1283–90.
Article
9. Rim CH, Cho WK, Lee JH, Kim YS, Suh YG, Kim KH, et al. Role of local treatment for oligometastasis: a comparability-based meta-analysis. Cancer Res Treat. 2022; 54:953–69.
Article
10. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009; 25:1754–60.
Article
11. Cibulskis K, Lawrence MS, Carter SL, Sivachenko A, Jaffe D, Sougnez C, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 2013; 31:213–9.
12. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010; 20:1297–303.
13. Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol. 2011; 29:24–6.
Article
14. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010; 38:e164.
Article
15. Forbes SA, Beare D, Boutselakis H, Bamford S, Bindal N, Tate J, et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res. 2017; 45:D777–83.
Article
16. Blokzijl F, Janssen R, van Boxtel R, Cuppen E. MutationalPatterns: comprehensive genome-wide analysis of mutational processes. Genome Med. 2018; 10:33.
Article
17. Chakravarty D, Gao J, Phillips SM, Kundra R, Zhang H, Wang J, et al. OncoKB: a precision oncology knowledge base. JCO Precis Oncol. 2017; 2017:PO.17.00011.
Article
18. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015; 348:69–74.
19. Subbiah V, Solit DB, Chan TA, Kurzrock R. The FDA approval of pembrolizumab for adult and pediatric patients with tumor mutational burden (TMB) ≥10: a decision centered on empowering patients and their physicians. Ann Oncol. 2020; 31:1115–8.
20. Teo MY, Seier K, Ostrovnaya I, Regazzi AM, Kania BE, Moran MM, et al. Alterations in DNA damage response and repair genes as potential marker of clinical benefit from PD-1/PD-L1 blockade in advanced urothelial cancers. J Clin Oncol. 2018; 36:1685–94.
21. Landrum MJ, Lee JM, Benson M, Brown G, Chao C, Chitipiralla S, et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 2016; 44:D862–8.
Article
22. Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, et al. A method and server for predicting damaging missense mutations. Nat Methods. 2010; 7:248–9.
23. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012; 16:284–7.
24. Alexandrov LB, Ju YS, Haase K, Van Loo P, Martincorena I, Nik-Zainal S, et al. Mutational signatures associated with tobacco smoking in human cancer. Science. 2016; 354:618–22.
Article
25. Helleday T, Eshtad S, Nik-Zainal S. Mechanisms underlying mutational signatures in human cancers. Nat Rev Genet. 2014; 15:585–98.
Article
26. Kim JH, Yoon S, Lee DH, Jang SJ, Chun SM, Kim SW. Real-world utility of next-generation sequencing for targeted gene analysis and its application to treatment in lung adenocarcinoma. Cancer Med. 2021; 10:3197–204.
Article
27. Huang RS, Harries L, Decker B, Hiemenz MC, Murugesan K, Creeden J, et al. Clinicopathologic and genomic landscape of non-small cell lung cancer brain metastases. Oncologist. 2022; 27:839–48.
28. Hellmann MD, Ciuleanu TE, Pluzanski A, Lee JS, Otterson GA, Audigier-Valette C, et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018; 378:2093–104.
Article
29. Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol. 2019; 30:44–56.
Article
30. Wang Z, Duan J, Cai S, Han M, Dong H, Zhao J, et al. Assessment of blood tumor mutational burden as a potential biomarker for immunotherapy in patients with non-small cell lung cancer with use of a next-generation sequencing cancer gene panel. JAMA Oncol. 2019; 5:696–702.
Article
31. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017; 9:34.
Article
32. Cremolini C, Antoniotti C, Rossini D, Lonardi S, Loupakis F, Pietrantonio F, et al. Upfront FOLFOXIRI plus bevacizumab and reintroduction after progression versus mFOLFOX6 plus bevacizumab followed by FOLFIRI plus bevacizumab in the treatment of patients with metastatic colorectal cancer (TRIBE2): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2020; 21:497–507.
33. Chen Z, Guo Y, Zhao D, Zou Q, Yu F, Zhang L, et al. Comprehensive analysis revealed that CDKN2A is a biomarker for immune infiltrates in multiple cancers. Front Cell Dev Biol. 2021; 9:808208.
Article
34. Offin M, Rizvi H, Tenet M, Ni A, Sanchez-Vega F, Li BT, et al. Tumor mutation burden and efficacy of EGFR-tyrosine kinase inhibitors in patients with EGFR-mutant lung cancers. Clin Cancer Res. 2019; 25:1063–9.
35. Chen H, Chong W, Wu Q, Yao Y, Mao M, Wang X. Association of LRP1B mutation with tumor mutation burden and outcomes in melanoma and non-small cell lung cancer patients treated with immune check-point blockades. Front Immunol. 2019; 10:1113.
Article
36. Schoenfeld AJ, Bandlamudi C, Lavery JA, Montecalvo J, Namakydoust A, Rizvi H, et al. The genomic landscape of SMARCA4 alterations and associations with outcomes in patients with lung cancer. Clin Cancer Res. 2020; 26:5701–8.
37. Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, et al. NCCN guidelines insights: non-small cell lung cancer, version 2.2021. J Natl Compr Canc Netw. 2021; 19:254–66.
38. Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, et al. Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018; 29:iv192–237.
Article
39. Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015; 161:1215–28.
Article
40. Egolf S, Zou J, Anderson A, Simpson CL, Aubert Y, Prouty S, et al. MLL4 mediates differentiation and tumor suppression through ferroptosis. Sci Adv. 2021; 7:eabj9141.
41. Whale AD, Colman L, Lensun L, Rogers HL, Shuttleworth SJ. Functional characterization of a novel somatic oncogenic mutation of PIK3CB. Signal Transduct Target Ther. 2017; 2:17063.
Article
42. Cancer Genome Atlas Research Network. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012; 489:519–25.
43. Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014; 511:543–50.
44. Frank R, Scheffler M, Merkelbach-Bruse S, Ihle MA, Kron A, Rauer M, et al. Clinical and pathological characteristics of KEAP1- and NFE2L2-mutated non-small cell lung carcinoma (NSCLC). Clin Cancer Res. 2018; 24:3087–96.
45. Blackford AN, Jackson SP. ATM, ATR, and DNA-PK: the trinity at the heart of the DNA damage response. Mol Cell. 2017; 66:801–17.
Article
46. Mayer IA, Abramson VG, Formisano L, Balko JM, Estrada MV, Sanders ME, et al. A phase Ib study of alpelisib (BYL719), a PI3Kalpha-specific inhibitor, with letrozole in ER+/HER2− metastatic breast cancer. Clin Cancer Res. 2017; 23:26–34.
47. Patnaik A, Appleman LJ, Tolcher AW, Papadopoulos KP, Beeram M, Rasco DW, et al. First-in-human phase I study of copanlisib (BAY 80–6946), an intravenous pan-class I phosphatidylinositol 3-kinase inhibitor, in patients with advanced solid tumors and non-Hodgkin’s lymphomas. Ann Oncol. 2016; 27:1928–40.
Article
48. Powles T, Lackner MR, Oudard S, Escudier B, Ralph C, Brown JE, et al. Randomized open-label phase II trial of apitolisib (GDC-0980), a novel inhibitor of the PI3K/mammalian target of rapamycin pathway, versus everolimus in patients with metastatic renal cell carcinoma. J Clin Oncol. 2016; 34:1660–8.
Article
49. Ma Z, Wang LZ, Cheng JT, Lam WST, Ma X, Xiang X, et al. Targeting hypoxia-inducible factor-1-mediated metastasis for cancer therapy. Antioxid Redox Signal. 2021; 34:1484–97.
50. Negrini S, Gorgoulis VG, Halazonetis TD. Genomic instability: an evolving hallmark of cancer. Nat Rev Mol Cell Biol. 2010; 11:220–8.
51. Zhao S, Zhang Z, Zhan J, Zhao X, Chen X, Xiao L, et al. Utility of comprehensive genomic profiling in directing treatment and improving patient outcomes in advanced non-small cell lung cancer. BMC Med. 2021; 19:223.
52. Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, et al. KRAS(G12C) inhibition with sotorasib in advanced solid tumors. N Engl J Med. 2020; 383:1207–17.
Article
53. Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, et al. Sotorasib for lung cancers with KRAS p.G12C mutation. N Engl J Med. 2021; 384:2371–81.
Article
54. Alborelli I, Leonards K, Rothschild SI, Leuenberger LP, Savic Prince S, Mertz KD, et al. Tumor mutational burden assessed by targeted NGS predicts clinical benefit from immune checkpoint inhibitors in non-small cell lung cancer. J Pathol. 2020; 250:19–29.
Article
55. Ricciuti B, Recondo G, Spurr LF, Li YY, Lamberti G, Venkatraman D, et al. Impact of DNA damage response and repair (DDR) gene mutations on efficacy of PD-(L)1 immune checkpoint inhibition in non-small cell lung cancer. Clin Cancer Res. 2020; 26:4135–42.
Article
56. Dong ZY, Zhong WZ, Zhang XC, Su J, Xie Z, Liu SY, et al. Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clin Cancer Res. 2017; 23:3012–24.
57. Wang C, Liang H, Lin C, Li F, Xie G, Qiao S, et al. Molecular subtyping and prognostic assessment based on tumor mutation burden in patients with lung adenocarcinomas. Int J Mol Sci. 2019; 20:4251.
58. Wang S, Zhang J, He Z, Wu K, Liu XS. The predictive power of tumor mutational burden in lung cancer immunotherapy response is influenced by patients’ sex. Int J Cancer. 2019; 145:2840–9.
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr